Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Laura Perani is active.

Publication


Featured researches published by Laura Perani.


Nature | 2006

Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs.

Maurilio Sampaolesi; Stéphane Blot; Giuseppe D'Antona; Nicolas Granger; Rossana Tonlorenzi; Anna Innocenzi; Paolo Mognol; Jean-Laurent Thibaud; Beatriz G. Gálvez; Inès Barthélémy; Laura Perani; Sara Mantero; Maria Guttinger; Orietta Pansarasa; Chiara Rinaldi; M. Gabriella Cusella De Angelis; Yvan Torrente; Claudio Bordignon; Roberto Bottinelli; Giulio Cossu

Duchenne muscular dystrophy remains an untreatable genetic disease that severely limits motility and life expectancy in affected children. The only animal model specifically reproducing the alterations in the dystrophin gene and the full spectrum of human pathology is the golden retriever dog model. Affected animals present a single mutation in intron 6, resulting in complete absence of the dystrophin protein, and early and severe muscle degeneration with nearly complete loss of motility and walking ability. Death usually occurs at about 1 year of age as a result of failure of respiratory muscles. Here we report that intra-arterial delivery of wild-type canine mesoangioblasts (vessel-associated stem cells) results in an extensive recovery of dystrophin expression, normal muscle morphology and function (confirmed by measurement of contraction force on single fibres). The outcome is a remarkable clinical amelioration and preservation of active motility. These data qualify mesoangioblasts as candidates for future stem cell therapy for Duchenne patients.


Science Translational Medicine | 2012

Transplantation of Genetically Corrected Human iPSC-Derived Progenitors in Mice with Limb-Girdle Muscular Dystrophy

Francesco Saverio Tedesco; Mattia F M Gerli; Laura Perani; Sara Benedetti; Federica Ungaro; Marco Cassano; Stefania Antonini; Enrico Tagliafico; Valentina Artusi; Emanuela Longa; Rossana Tonlorenzi; Martina Ragazzi; Giorgia Calderazzi; Hidetoshi Hoshiya; Ornella Cappellari; Marina Mora; Benedikt Schoser; Peter Schneiderat; Mitsuo Oshimura; Roberto Bottinelli; Maurilio Sampaolesi; Yvan Torrente; Vania Broccoli; Giulio Cossu

Genetically corrected mesoangioblasts from human iPSCs derived from limb-girdle muscular dystrophy patients produce muscle fibers expressing the therapeutic gene in a mouse model of the disease. Muscle Progenitors Find Their Way Home Muscular dystrophies are genetic disorders primarily affecting skeletal muscle that result in greatly impaired mobility and, in severe cases, respiratory and cardiac dysfunction. There is no effective treatment, although several new approaches are entering clinical testing including cell therapy. Cell therapy aims to replace lost muscle fibers by transplanting healthy donor muscle progenitor cells or cells from dystrophic patients that have been genetically corrected in vitro. Mesoangioblasts are progenitor cells from blood vessel walls that have shown potential as a cell therapy in animal models of muscular dystrophy. In a new study, Tedesco et al. explore whether genetically corrected mesoangioblasts from patients with limb-girdle muscular dystrophy 2D (LGMD2D) have potential as an autologous cell therapy to treat this disease. The authors quickly found that they could not derive a sufficient number of mesoangioblasts from LGMD2D patients because the muscles of the patients were depleted of these progenitor cells. To overcome this problem, the authors reprogrammed fibroblasts or myoblasts from the LGMD2D patients to obtain human induced pluripotent stem cells (iPSCs) and induced them to differentiate into mesoangioblast-like cells that were then genetically corrected in vitro using a viral vector expressing the defective gene SGCA, which encodes α-sarcoglycan. After intramuscular or intra-arterial injection of these genetically corrected, iPSC-derived mesoangioblasts into mice with LGMD2D (immune-deficient Sgca-null mice), the cells homed to damaged mouse skeletal muscle, engrafted, and formed muscle fibers expressing α-sarcoglycan. Using mouse iPSC-derived mesoangioblasts, the researchers showed that the transplanted engrafted cells imbued muscle with greater strength and enabled the dystrophic mice to run for longer on a treadmill than dystrophic mice that did not receive the cells. This strategy offers the advantage of being able to produce unlimited numbers of genetically corrected progenitor cells, which perhaps could be used in the future as cell therapy for treating LGMD2D and other forms of muscular dystrophy. Mesoangioblasts are stem/progenitor cells derived from a subset of pericytes found in muscle that express alkaline phosphatase. They have been shown to ameliorate the disease phenotypes of different animal models of muscular dystrophy and are now undergoing clinical testing in children affected by Duchenne’s muscular dystrophy. Here, we show that patients with a related disease, limb-girdle muscular dystrophy 2D (LGMD2D), which is caused by mutations in the gene encoding α-sarcoglycan, have reduced numbers of this pericyte subset and thus produce too few mesoangioblasts for use in autologous cell therapy. Hence, we reprogrammed fibroblasts and myoblasts from LGMD2D patients to generate human induced pluripotent stem cells (iPSCs) and developed a protocol for the derivation of mesoangioblast-like cells from these iPSCs. The iPSC-derived mesoangioblasts were expanded and genetically corrected in vitro with a lentiviral vector carrying the gene encoding human α-sarcoglycan and a promoter that would ensure expression only in striated muscle. When these genetically corrected human iPSC-derived mesoangioblasts were transplanted into α-sarcoglycan–null immunodeficient mice, they generated muscle fibers that expressed α-sarcoglycan. Finally, transplantation of mouse iPSC-derived mesoangioblasts into α-sarcoglycan–null immunodeficient mice resulted in functional amelioration of the dystrophic phenotype and restoration of the depleted progenitors. These findings suggest that transplantation of genetically corrected mesoangioblast-like cells generated from iPSCs from LGMD2D patients may be useful for treating this type of muscular dystrophy and perhaps other forms of muscular dystrophy as well.


Cell | 2010

Nfix Regulates Fetal-Specific Transcription in Developing Skeletal Muscle

Graziella Messina; Stefano Biressi; Stefania Monteverde; Alessandro Magli; Marco Cassano; Laura Perani; Elena Roncaglia; Enrico Tagliafico; Linda M. Starnes; Christine E. Campbell; Milena Grossi; David J. Goldhamer; Richard M. Gronostajski; Giulio Cossu

Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.


Cell Death and Disease | 2010

Partial dysferlin reconstitution by adult murine mesoangioblasts is sufficient for full functional recovery in a murine model of dysferlinopathy

Jordi Díaz-Manera; T. Touvier; Arianna Dellavalle; Rossana Tonlorenzi; Francesco Saverio Tedesco; Graziella Messina; M..A. Meregalli; C. Navarro; Laura Perani; C. Bonfanti; I. Illa; Yvan Torrente; Giulio Cossu

Dysferlin deficiency leads to a peculiar form of muscular dystrophy due to a defect in sarcolemma repair and currently lacks a therapy. We developed a cell therapy protocol with wild-type adult murine mesoangioblasts. These cells differentiate with high efficiency into skeletal muscle in vitro but differ from satellite cells because they do not express Pax7. After intramuscular or intra-arterial administration to SCID/BlAJ mice, a novel model of dysferlinopathy, wild-type mesoangioblasts efficiently colonized dystrophic muscles and partially restored dysferlin expression. Nevertheless, functional assays performed on isolated single fibers from transplanted muscles showed a normal repairing ability of the membrane after laser-induced lesions; this result, which reflects gene correction of an enzymatic rather than a structural deficit, suggests that this myopathy may be easier to treat with cell or gene therapy than other forms of muscular dystrophies.


Nature | 2013

Corrigendum: Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs

Maurilio Sampaolesi; Stéphane Blot; Giuseppe D'Antona; Nicolas Granger; Rossana Tonlorenzi; Anna Innocenzi; Paolo Mognol; Jean-Lauren Thibaud; Beatriz G. Gálvez; Inès Barthélémy; Laura Perani; Sara Mantero; Maria Guttinger; Orietta Pansarasa; Chiara Rinaldi; M. Gabriella Cusella De Angelis; Yvan Torrente; Claudio Bordignon; Roberto Bottinelli; Giulio Cossu

This corrects the article DOI: 10.1038/nature05282


Scientific Reports | 2016

Adaptive immunity against gut microbiota enhances apoE-mediated immune regulation and reduces atherosclerosis and western-diet-related inflammation

Diego Saita; Roberto Ferrarese; Chiara Foglieni; Antonio Esposito; Tamara Canu; Laura Perani; Elisa Rita Ceresola; Laura Visconti; Roberto Burioni; Massimo Clementi; Filippo Canducci

Common features of immune-metabolic and inflammatory diseases such as metabolic syndrome, diabetes, obesity and cardiovascular diseases are an altered gut microbiota composition and a systemic pro-inflammatory state. We demonstrate that active immunization against the outer membrane protein of bacteria present in the gut enhances local and systemic immune control via apoE-mediated immune-modulation. Reduction of western-diet-associated inflammation was obtained for more than eighteen weeks after immunization. Immunized mice had reduced serum cytokine levels, reduced insulin and fasting glucose concentrations; and gene expression in both liver and visceral adipose tissue confirmed a reduced inflammatory steady-state after immunization. Moreover, both gut and atherosclerotic plaques of immunized mice showed reduced inflammatory cells and an increased M2 macrophage fraction. These results suggest that adaptive responses directed against microbes present in our microbiota have systemic beneficial consequences and demonstrate the key role of apoE in this mechanism that could be exploited to treat immune-metabolic diseases.


Developmental Biology | 2012

Noggin recruits mesoderm progenitors from the dorsal aorta to a skeletal myogenic fate

Gonzalo Ugarte; Ornella Cappellari; Laura Perani; Anna Pistocchi; Giulio Cossu

Embryonic mesoangioblasts are the in vitro counterpart of vessel-associated progenitors, able to differentiate into different mesoderm cell types. To investigate signals recruiting these progenitors to a skeletal myogenic fate, we developed an in vitro assay, based upon co-culture of E11.5 dorsal aorta (from MLC3 F-nLacZ transgenic embryos, expressing nuclear beta galactosidase only in striated muscle) with differentiating C2C12 or primary myoblasts. Under these conditions muscle differentiation from cells originating from the vessel can be quantified by counting the number of beta gal + nuclei. Results indicated that Noggin (but not Follistatin, Chordin or Gremlin) stimulates while BMP2/4 inhibits myogenesis from dorsal aorta progenitors; neutralizing antibodies and shRNA greatly reduce these effects. In contrast, TGF-β1, VEGF, Wnt7A, Wnt3A, bFGF, PDGF-BB and IGF1 have no effect. Sorting experiments indicated that the majority of these myogenic progenitors express the pericyte marker NG2. Moreover they are abundant in the thoracic segment at E10.5 and in the iliac bifurcation at E11.5 suggesting the occurrence of a cranio-caudal wave of competent cells along the aorta. BMP2 is expressed in the dorsal aorta and Noggin in newly formed muscle fibers suggesting that these two tissues compete to recruit mesoderm cells to a myogenic or to a perithelial fate in the developing fetal muscle.


Scientific Reports | 2016

Human malignant mesothelioma is recapitulated in immunocompetent BALB/c mice injected with murine AB cells

Rosanna Mezzapelle; Eltjona Rrapaj; Elena Gatti; Chiara Ceriotti; Francesco De Marchis; Alessandro Preti; Antonello E. Spinelli; Laura Perani; Massimo Venturini; Silvia Valtorta; Rosa Maria Moresco; Lorenza Pecciarini; Claudio Doglioni; Michela Frenquelli; Luca Crippa; Camilla Recordati; Eugenio Scanziani; Hilda de Vries; Anton Berns; Roberta Frapolli; Renzo Boldorini; Maurizio D’Incalci; Marco Bianchi; Massimo P. Crippa

Malignant Mesothelioma is a highly aggressive cancer, which is difficult to diagnose and treat. Here we describe the molecular, cellular and morphological characterization of a syngeneic system consisting of murine AB1, AB12 and AB22 mesothelioma cells injected in immunocompetent BALB/c mice, which allows the study of the interplay of tumor cells with the immune system. Murine mesothelioma cells, like human ones, respond to exogenous High Mobility Group Box 1 protein, a Damage-Associated Molecular Pattern that acts as a chemoattractant for leukocytes and as a proinflammatory mediator. The tumors derived from AB cells are morphologically and histologically similar to human MM tumors, and respond to treatments used for MM patients. Our system largely recapitulates human mesothelioma, and we advocate its use for the study of MM development and treatment.


Advanced Functional Materials | 2017

Glycine N-Methylation in NGR-Tagged Nanocarriers Prevents Isoaspartate Formation and Integrin Binding without Impairing CD13 Recognition and Tumor Homing

Angelo Corti; Anna Gasparri; Michela Ghitti; Angelina Sacchi; Francesco Sudati; Martina Fiocchi; Valentina Buttiglione; Laura Perani; Alessandro Gori; Silvia Valtorta; Rosa Maria Moresco; Fabio Pastorino; Mirco Ponzoni; Giovanna Musco; Flavio Curnis

NGR (asparagine-glycine-arginine) is a tumor vasculature-homing peptide motif widely used for the functionalization of drugs, nanomaterials and imaging compounds for cancer treatment and diagnosis. Unfortunately, this motif has a strong propensity to undergo rapid deamidation. This reaction, which converts NGR into isoDGR, is associated with receptor switching from CD13 to integrins, with potentially important manufacturing, pharmacological and toxicological implications. It is found that glycine N-methylation of NGR-tagged nanocarriers completely prevents asparagine deamidation without impairing CD13 recognition. Studies in animal models have shown that the methylated NGR motif can be exploited for delivering radiolabeled compounds and nanocarriers, such as tumor necrosis factor-α (TNF)-bearing nanogold and liposomal doxorubicin, to tumors with improved selectivity. These findings suggest that this NGR derivative is a stable and efficient tumor-homing ligand that can be used for delivering functional nanomaterials to tumor vasculature.


Oncotarget | 2016

Regulation of tumor growth by circulating full-length chromogranin A

Flavio Curnis; Alice Dallatomasina; Mimma Bianco; Anna Gasparri; Angelina Sacchi; Barbara Colombo; Martina Fiocchi; Laura Perani; Massimo Venturini; Carlo Tacchetti; Suvajit Sen; Ricardo Borges; Eleonora Dondossola; Antonio Esposito; Sushil K. Mahata; Angelo Corti

Chromogranin A (CgA), a neuroendocrine secretory protein, and its fragments are present in variable amounts in the blood of normal subjects and cancer patients. We investigated whether circulating CgA has a regulatory function in tumor biology and progression. Systemic administration of full-length CgA, but not of fragments lacking the C-terminal region, could reduce tumor growth in murine models of fibrosarcoma, mammary adenocarcinoma, Lewis lung carcinoma, and primary and metastatic melanoma, with U-shaped dose-response curves. Tumor growth inhibition was associated with reduction of microvessel density and blood flow in neoplastic tissues. Neutralization of endogenous CgA with antibodies against its C-terminal region (residues 410-439) promoted tumor growth. Structure-function studies showed that the C-terminal region of CgA contains a bioactive site and that cleavage of this region causes a marked loss of anti-angiogenic and anti-tumor potency. Mechanistic studies showed that full-length CgA could induce, with a U-shaped dose-response curve, the production of protease nexin-1 in endothelial cells, a serine protease inhibitor endowed of anti-angiogenic activity. Gene silencing or neutralization of protease nexin-1 with specific antibodies abolished both anti-angiogenic and anti-tumor effects of CgA. These results suggest that circulating full-length CgA is an important inhibitor of angiogenesis and tumor growth, and that cleavage of its C-terminal region markedly reduces its activity. Pathophysiological changes in CgA blood levels and/or its fragmentation might regulate disease progression in cancer patients.

Collaboration


Dive into the Laura Perani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rossana Tonlorenzi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Yvan Torrente

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Maurilio Sampaolesi

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Anna Innocenzi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beatriz G. Gálvez

Centro Nacional de Investigaciones Cardiovasculares

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge